Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.443
Filter
1.
Mol Vis ; 30: 49-57, 2024.
Article in English | MEDLINE | ID: mdl-38586605

ABSTRACT

RPGR pathogenic variants are the major cause of X-linked retinitis pigmentosa. Here, we report the results from 1,033 clinical DNA tests that included sequencing of RPGR. A total of 184 RPGR variants were identified: 78 pathogenic or likely pathogenic, 14 uncertain, and 92 likely benign or benign. Among the pathogenic and likely pathogenic variants, 23 were novel, and most were frameshift or nonsense mutations (87%) and enriched (67%) in RPGR exon 15 (ORF15). Identical pathogenic variants found in different families were largely on different haplotype backgrounds, indicating relatively frequent, recurrent RPGR mutations. None of the 16 mother/affected son pairs showed de novo mutations; all 16 mothers were heterozygous for the pathogenic variant. These last two observations support the occurrence of most RPGR mutations in the male germline.


Subject(s)
Eye Proteins , Retinitis Pigmentosa , Humans , Eye Proteins/genetics , Pedigree , Mutation , Frameshift Mutation , Vision Disorders , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
2.
Tomography ; 10(4): 480-492, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38668395

ABSTRACT

The aim of this paper is to investigate whether a multifractal analysis can be applied to study choroidal blood vessels and help ophthalmologists in the early diagnosis of retinitis pigmentosa (RP). In a case study, we used spectral domain optical coherence tomography (SDOCT), which is a noninvasive and highly sensitive imaging technique of the retina and choroid. The image of a choroidal branching pattern can be regarded as a multifractal. Therefore, we calculated the generalized Renyi point-centered dimensions, which are considered a measure of the inhomogeneity of data, to prove that it increases in patients with RP as compared to those in the control group.


Subject(s)
Choroid , Retinitis Pigmentosa , Tomography, Optical Coherence , Adult , Female , Humans , Male , Middle Aged , Choroid/diagnostic imaging , Choroid/pathology , Fractals , Retinitis Pigmentosa/diagnostic imaging , Retinitis Pigmentosa/pathology , Tomography, Optical Coherence/methods
3.
Genes (Basel) ; 15(4)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674450

ABSTRACT

Retinitis pigmentosa is a group of genetically determined retinal dystrophies characterized by primary photoreceptor apoptosis and can occur in isolated or syndromic conditions. This study reviewed the clinical data of 15 patients with syndromic retinitis pigmentosa from a Rare Disease Reference Center in Brazil and the results of their next-generation sequencing tests. Five males and ten females participated, with the mean ages for ocular disease onset, fundoscopic diagnosis, and molecular evaluation being 9, 19, and 29 years, respectively. Bardet-Biedl syndrome (n = 5) and Usher syndrome (n = 3) were the most frequent diagnoses, followed by other rare conditions. Among the patients, fourteen completed molecular studies, with three negative results and eleven revealing findings in known genes, including novel variants in MKKS (c.432_435del, p.Phe144Leufs*14), USH2A (c.(7301+1_7302-1)_(9369+1_9370-1)del), and CEP250 (c.5383dup, p.Glu1795Glyfs*13, and c.5050del, p.Asp1684Thrfs*9). Except for Kearn-Sayre, all presented an autosomal recessive inheritance pattern with 64% homozygosity results. The long gap between symptom onset and diagnosis highlights the diagnostic challenges faced by the patients. This study reaffirms the clinical heterogeneity of syndromic retinitis pigmentosa and underscores the pivotal role of molecular analysis in advancing our understanding of these diseases.


Subject(s)
Retinitis Pigmentosa , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/diagnosis , Male , Female , Adult , Adolescent , Child , Young Adult , Usher Syndromes/genetics , Usher Syndromes/pathology , Usher Syndromes/diagnosis , Brazil/epidemiology , Middle Aged , High-Throughput Nucleotide Sequencing , Bardet-Biedl Syndrome/genetics , Mutation
4.
Neurosci Lett ; 830: 137778, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38621504

ABSTRACT

The endoplasmic reticulum (ER) plays an indispensable role in cellular processes, including maintenance of calcium homeostasis, and protein folding, synthesized and processing. Disruptions in these processes leading to ER stress and the accumulation of misfolded proteins can instigate the unfolded protein response (UPR), culminating in either restoration of balanced proteostasis or apoptosis. A key player in this intricate balance is CLCC1, an ER-resident chloride channel, whose essential role extends to retinal development, regulation of ER stress, and UPR. The importance of CLCC1 is further underscored by its interaction with proteins localized to mitochondria-associated endoplasmic reticulum membranes (MAMs), where it participates in UPR induction by MAM proteins. In previous research, we identified a p.(Asp25Glu) pathogenic CLCC1 variant associated with retinitis pigmentosa (RP) (CLCC1 hg38 NC_000001.11; NM_001048210.3, c.75C > A; UniprotKB Q96S66). In attempt to decipher the impact of this variant function, we leveraged liquid chromatography-mass spectrometry (LC-MS) to identify likely CLCC1-interacting proteins. We discovered that the CLCC1 interactome is substantially composed of proteins that localize to ER compartments and that the Asp25Glu variant results in noticeable loss and gain of specific protein interactors. Intriguingly, the analysis suggests that the CLCC1Asp25Glu mutant protein exhibits a propensity for increased interactions with cytoplasmic proteins compared to its wild-type counterpart. To corroborate our LC-MS data, we further scrutinized two novel CLCC1 interactors, Calnexin and SigmaR1, chaperone proteins that localize to the ER and MAMs. Through microscopy, we demonstrate that CLCC1 co-localizes with both proteins, thereby validating our initial findings. Moreover, our results reveal that CLCC1 co-localizes with SigmaR1 not merely at the ER, but also at MAMs. These findings reinforce the notion of CLCC1 interacting with MAM proteins at the ER-mitochondria interface, setting the stage for further exploration into how these interactions impact ER or mitochondria function and lead to retinal degenerative disease when impaired.


Subject(s)
Endoplasmic Reticulum , Receptors, sigma , Sigma-1 Receptor , Humans , Endoplasmic Reticulum/metabolism , HEK293 Cells , Mitochondria/metabolism , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Receptors, sigma/metabolism , Receptors, sigma/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Unfolded Protein Response
5.
Int J Biol Macromol ; 268(Pt 2): 131671, 2024 May.
Article in English | MEDLINE | ID: mdl-38641272

ABSTRACT

In this study, N-Methyl-N-nitrosourea (MNU) was intraperitoneally injected to construct a mouse retinitis pigmentosa (RP) model to evaluate the protective effect of chitosan and ß-carotene on RP. The results demonstrated that chitosan synergized with ß-carotene significantly reduced retinal histopathological structural damage in RP mice. The co-treatment group of ß-carotene and chitosan restored the retinal thickness and outer nuclear layer thickness better than the group treated with the two alone, and the thickness reached the normal level. The content of ß-carotene and retinoids in the liver of chitosan and ß-carotene co-treated group increased by 46.75 % and 20.69 %, respectively, compared to the ß-carotene group. Chitosan and ß-carotene supplement suppressed the expressions of Bax, Calpain2, Caspase3, NF-κB, TNF-α, IL-6, and IL-1ß, and promoted the up-regulation of Bcl2. Chitosan and ß-carotene interventions remarkably contributed to the content of SCFAs and enhanced the abundance of Ruminococcaceae, Rikenellaceae, Odoribacteraceae and Helicobacteraceae. Correlation analysis demonstrated a strong association between gut microbiota and improvement in retinitis pigmentosa. This study will provide a reference for the study of the gut-eye axis.


Subject(s)
Chitosan , Methylnitrosourea , Retinitis Pigmentosa , beta Carotene , Animals , beta Carotene/pharmacology , Chitosan/pharmacology , Chitosan/chemistry , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Mice , Drug Synergism , Retina/drug effects , Retina/metabolism , Retina/pathology , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Male , Retinoids/pharmacology , Liver/drug effects , Liver/pathology , Liver/metabolism
6.
FASEB J ; 38(8): e23606, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648465

ABSTRACT

Rhodopsin mislocalization encompasses various blind conditions. Rhodopsin mislocalization is the primary factor leading to rod photoreceptor dysfunction and degeneration in autosomal dominant retinitis pigmentosa (adRP) caused by class I mutations. In this study, we report a new knock-in mouse model that harbors a class I Q344X mutation in the endogenous rhodopsin gene, which causes rod photoreceptor degeneration in an autosomal dominant pattern. In RhoQ344X/+ mice, mRNA transcripts from the wild-type (Rho) and RhoQ344X mutant rhodopsin alleles are expressed at equal levels. However, the amount of RHOQ344X mutant protein is 2.7 times lower than that of wild-type rhodopsin, a finding consistent with the rapid degradation of the mutant protein. Immunofluorescence microscopy indicates that RHOQ344X is mislocalized to the inner segment and outer nuclear layers of rod photoreceptors in both RhoQ344X/+ and RhoQ344X/Q344X mice, confirming the essential role of the C-terminal VxPx motif in promoting OS delivery of rhodopsin. The mislocalization of RHOQ344X is associated with the concurrent mislocalization of wild-type rhodopsin in RhoQ344X/+ mice. To understand the global changes in proteostasis, we conducted quantitative proteomics analysis and found attenuated expression of rod-specific OS membrane proteins accompanying reduced expression of ciliopathy causative gene products, including constituents of BBSome and axonemal dynein subunit. Those studies unveil a novel negative feedback regulation involving ciliopathy-associated proteins. In this process, a defect in the trafficking signal leads to a reduced quantity of the trafficking apparatus, culminating in a widespread reduction in the transport of ciliary proteins.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Retinal Rod Photoreceptor Cells , Retinitis Pigmentosa , Rhodopsin , Animals , Rhodopsin/metabolism , Rhodopsin/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Cilia/metabolism , Cilia/pathology
7.
Stem Cell Res ; 77: 103390, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38507880

ABSTRACT

Retinitis pigmentosa (RP) is a group of genetically heterogeneous retinopathy resulting in irreversible loss of vision. Mutations in RAX2 gene has been related to RP with mechanisms unclear. Here, we generated a human induced pluripotent stem cell (iPSC) line from peripheral blood mononuclear cells of a RP patient carrying c.77C > T mutation in RAX2 gene. This cell line was induced by integration-free episomal vectors and validated for pluripotency and differentiation capacity, which may serve as a model to study the role of RAX2 in RP pathogenesis.


Subject(s)
Homeodomain Proteins , Induced Pluripotent Stem Cells , Mutation , Retinitis Pigmentosa , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Induced Pluripotent Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cell Line , Cell Differentiation , Eye Proteins/genetics , Eye Proteins/metabolism , Male , Transcription Factors
8.
Gene ; 905: 148237, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38310983

ABSTRACT

Approximately a quarter of Retinitis Pigmentosa (RP) is caused by mutations in transport-related genes in cilia. IFT27 (Intraflagellar Transport 27), a core component of the ciliary intraflagellar transport (IFT) system, has been implicated as a significant pathogenic gene in RP. The pathogenic mechanisms and subsequent pathology related to IFT27 mutations in RP are largely obscure. Here, we utilized TALEN technology to create an ift27 knockout (ift27-/-) zebrafish model. Electroretinography (ERG) detection showed impaired vision in this model. Histopathological examinations disclosed that ift27 mutations cause progressive degeneration of photoreceptors in zebrafish, and this degeneration was late-onset. Immunofluorescence labeling of outer segments showed that rods degenerated before cones, aligning with the conventional characterization of RP. In cultured human retinal pigment epithelial cells, we found that IFT27 was involved in maintaining ciliary morphology. Furthermore, decreased IFT27 expression resulted in the inhibition of the Hedgehog (Hh) signaling pathway, including decreased expression of key factors in the Hh pathway and abnormal localization of the ciliary mediator Gli2. In summary, we generated an ift27-/- zebrafish line with retinal degeneration which mimicked the symptoms of RP patients, highlighting IFT27's integral role in the long-term maintenance of cilia via the Hh signaling pathway. This work may furnish new insights into the treatment or delay of RP caused by IFT27 mutations.


Subject(s)
Retinitis Pigmentosa , Zebrafish Proteins , Zebrafish , Animals , Humans , Biological Transport , Cilia/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
9.
Stem Cell Res ; 76: 103334, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38340451

ABSTRACT

Retinitis pigmentosa (RP) is the most common inherited retinal diseases, characterized by photoreceptor cell death and retinal pigment epithelial atrophy. Mutations in cyclic nucleotide gated channel subunit alpha 1 (CNGA1) have been reported to cause retinitis pigmentosa. Here, we established the human induced pluripotent stem cell line (iPSC) SJTUGHi002-A, generated from peripheral blood mononuclear cells of a 36-year-old male RP patient, who carried a homozygous frameshift variant in CNGA1 gene (c.265delC; p.L89Ffs*4). The cell line can serve as a patient-derived disease model for exploring the pathogenesis and drug development of CNGA1-RP.


Subject(s)
Induced Pluripotent Stem Cells , Retinitis Pigmentosa , Adult , Humans , Male , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Mutation , Retinitis Pigmentosa/pathology
10.
Exp Eye Res ; 240: 109826, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38340947

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinal disorder characterized by the degeneration of photoreceptors. RhoP23H/+ mice, which carry a Pro23His mutation in the RHODOPSIN (Rho) gene, are one of the most studied animal models for RP. However, except for the photoreceptors, other retinal neural cells have not been fully investigated in this model. Here, we record the temporal changes of the retina by optical coherence tomography (OCT) imaging of the RhoP23H/+ mice, from early to mid-phase of retinal degeneration. Based on thickness analysis, we identified a natural retinal thickness adaption in wild-type mice during early adulthood and observed morphological compensation of the inner retina layer to photoreceptor degeneration in the RhoP23H/+ mice, primarily on the inner nuclear layer (INL). RhoP23H/+ mice findings were further validated via: histology showing the negative correlation of INL and ONL thicknesses; as well as electroretinogram (ERG) showing an increased b-wave to a-wave ratio. These results unravel the sequential morphologic events in this model and suggest a better understanding of retinal degeneration of RP for future studies.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Mice , Animals , Retinal Degeneration/diagnosis , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Rhodopsin/genetics , Retina/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Electroretinography , Disease Models, Animal
11.
Cell ; 187(6): 1387-1401.e13, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38412859

ABSTRACT

The Crumbs homolog 1 (CRB1) gene is associated with retinal degeneration, most commonly Leber congenital amaurosis (LCA) and retinitis pigmentosa (RP). Here, we demonstrate that murine retinas bearing the Rd8 mutation of Crb1 are characterized by the presence of intralesional bacteria. While normal CRB1 expression was enriched in the apical junctional complexes of retinal pigment epithelium and colonic enterocytes, Crb1 mutations dampened its expression at both sites. Consequent impairment of the outer blood retinal barrier and colonic intestinal epithelial barrier in Rd8 mice led to the translocation of intestinal bacteria from the lower gastrointestinal (GI) tract to the retina, resulting in secondary retinal degeneration. Either the depletion of bacteria systemically or the reintroduction of normal Crb1 expression colonically rescued Rd8-mutation-associated retinal degeneration without reversing the retinal barrier breach. Our data elucidate the pathogenesis of Crb1-mutation-associated retinal degenerations and suggest that antimicrobial agents have the potential to treat this devastating blinding disease.


Subject(s)
Nerve Tissue Proteins , Retinal Degeneration , Animals , Mice , Bacterial Translocation , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
12.
Ophthalmic Genet ; 45(2): 113-119, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38186350

ABSTRACT

BACKGROUND: PHARC syndrome (MIM:612674) is a rare neurodegenerative disorder characterized by demyelinating polyneuropathy, hearing loss, ataxia, retinitis pigmentosa, and cataracts (PHARC). The syndrome is caused by mutations in the ABHD12 gene, which encodes αß-hydrolase domain-containing protein 12 related to endocannabinoid metabolism. PHARC syndrome is one of the rare diseases; so far, only 51 patients have been reported in the literature. METHODS: We evaluated the 25-year-old male patient referred to us due to vision loss, cataracts, and hearing loss. Ophthalmological examinations and genetic analyses were performed using targeted next-generation sequencing. RESULTS: In the genetic analysis, the patient was diagnosed with PHARC syndrome by detecting homozygous (NM_001042472.3): c.871del (p.Tyr291IlefsTer28) novel pathogenic variation in the ABHD12 gene. Following the molecular diagnosis, he was referred to the neurology department for reverse phenotyping and sensorimotor demyelinating polyneuropathy was detected in the neurological evaluation. CONCLUSIONS: In this study, we report a novel variation in ABHD12 gene in the first Turkish-origin PHARC patient. We present this study to contribute genotype-phenotype correlation of PHARC syndrome and emphasize the importance of molecular genetic diagnosis in order to determine the appropriate clinical approach. This report is essential for expanding the phenotypic spectrum in different populations and understanding the genotype-phenotype correlation of PHARC syndrome via novel pathogenic variation in the ABHD12 gene.


Subject(s)
Ataxia , Cataract , Hearing Loss , Polyneuropathies , Retinitis Pigmentosa , Male , Humans , Adult , Phenotype , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Mutation , Syndrome , Cataract/diagnosis , Cataract/genetics , Polyneuropathies/diagnosis , Polyneuropathies/genetics , Polyneuropathies/pathology , Pedigree , Monoacylglycerol Lipases/genetics
13.
Ophthalmic Genet ; 45(1): 63-71, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37160720

ABSTRACT

PURPOSE: Mutations in the SCAPER gene have previously been reported to be a rare cause of syndromic and non-syndromic autosomal recessive retinitis pigmentosa (RP). We report a case of syndromic RP caused by a frameshift heterozygous mutation in SCAPER. Our case has a relatively mild ocular phenotype with the presence of cone involvement noted on full field electroretinogram (ffERG) without impacting central or color vision. MATERIALS AND METHODS: A 17-year-old male presented with progressive nyctalopia in both eyes. He underwent ophthalmic examination and multimodal imaging. A complete retinal degeneration panel consisting of 322 genes was used to screen for molecular causes of retinal dystrophy in this patient along with family segregation analysis. RESULTS: Fundus examination of the proband revealed mild RP phenotype with waxy pallor of optic discs, attenuated retinal arterioles, and single bone spicule like pigmentary change in the mid-periphery bilaterally. Multimodal imaging and ffERG demonstrated a picture of RP with cone dysfunction without impacting central or color vision bilaterally. Examined family members were found to be normal. The proband was found to be heterozygous for two novel frameshift pathogenic variants in SCAPER c.3781del, p. (Val1261Serfs*26), c.868_869del, p. (Glu290Serfs*7) both leading to predicted premature termination. The family members tested were found to be heterozygous for SCAPER c.868_869del, p. (Glu290Serfs*7) pathogenic variant confirming their carrier status. CONCLUSION: We report a case of a syndromic RP of previously unreported ocular phenotype associated with SCAPER pathogenic variant, which will add to the phenotypic spectrum of retinopathy and systemic features associated with pathogenic variants in SCAPER.


Subject(s)
Retinal Dystrophies , Retinitis Pigmentosa , Male , Humans , Adolescent , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Frameshift Mutation , Mutation , Phenotype , Retinal Cone Photoreceptor Cells/pathology , Pedigree , Carrier Proteins/genetics
14.
Int J Mol Sci ; 24(24)2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38139011

ABSTRACT

Many retinal degenerative diseases result in vision impairment or permanent blindness due to photoreceptor loss or dysfunction. It has been observed that Pde6brd1 mice (rd1), which carry a spontaneous nonsense mutation in the pde6b gene, have a strong phenotypic similarity to patients suffering from autosomal recessive retinitis pigmentosa. In this study, we present a novel mouse model of retinitis pigmentosa generated through pde6b gene knockout using CRISPR/Cas9 technology. We compare this Pde6b-KO mouse model to the rd1 mouse model to gain insights into the progression of retinal degeneration. The functional assessment of the mouse retina and the tracking of degeneration dynamics were performed using electrophysiological methods, while retinal morphology was analyzed through histology techniques. Interestingly, the Pde6b-KO mouse model demonstrated a higher amplitude of photoresponse than the rd1 model of the same age. At postnatal day 12, the thickness of the photoreceptor layer in both mouse models did not significantly differ from that of control animals; however, by day 15, a substantial reduction was observed. Notably, the decline in the number of photoreceptors in the rd1 model occurred at a significantly faster rate. These findings suggest that the C3H background may play a significant role in the early stages of retinal degeneration.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Humans , Mice , Animals , Retinal Degeneration/pathology , Electroretinography , Mice, Inbred C3H , Retinitis Pigmentosa/pathology , Retina/pathology , Disease Models, Animal
15.
Stem Cell Res Ther ; 14(1): 340, 2023 11 27.
Article in English | MEDLINE | ID: mdl-38012786

ABSTRACT

Inherited retinal diseases (IRDs) can induce severe sight-threatening retinal degeneration and impose a considerable economic burden on patients and society, making efforts to cure blindness imperative. Transgenic animals mimicking human genetic diseases have long been used as a primary research tool to decipher the underlying pathogenesis, but there are still some obvious limitations. As an alternative strategy, patient-derived induced pluripotent stem cells (iPSCs), particularly three-dimensional (3D) organoid technology, are considered a promising platform for modeling different forms of IRDs, including retinitis pigmentosa, Leber congenital amaurosis, X-linked recessive retinoschisis, Batten disease, achromatopsia, and best vitelliform macular dystrophy. Here, this paper focuses on the status of patient-derived iPSCs and organoids in IRDs in recent years concerning disease modeling and therapeutic exploration, along with potential challenges for translating laboratory research to clinical application. Finally, the importance of human iPSCs and organoids in combination with emerging technologies such as multi-omics integration analysis, 3D bioprinting, or microfluidic chip platform are highlighted. Patient-derived retinal organoids may be a preferred choice for more accurately uncovering the mechanisms of human retinal diseases and will contribute to clinical practice.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Animals , Humans , Induced Pluripotent Stem Cells/pathology , Retina/pathology , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/pathology , Organoids
16.
Genes (Basel) ; 14(10)2023 10 17.
Article in English | MEDLINE | ID: mdl-37895299

ABSTRACT

The goal of the study was to explore the spectrum of pathogenic variants in the RPGR gene in a group of male Polish patients with a retinitis pigmentosa (RP) phenotype. A total of 45 male index patients, including twins, being members of 44 families, were screened for pathogenic variants in the RPGR gene via the direct sequencing of PCR-amplified genomic DNA and underwent a comprehensive ophthalmological examination in one center located in Poland. A total of two pathogenic and five likely pathogenic variants in eight patients (18%) were detected in the studied cohort. Of these, five variants were novel, and five disease-causing variants (71%) were identified within the ORF15 mutational hotspot of the RPGR gene. The median age of onset of the disease was 10 years (range 6-14 years), the median age during the examination was 30 years (range 20-47 years), and the median visual acuity was 0.4 (range 0.01-0.7). The majority of patients had middle constriction of the visual field and thinning of the central foveal thickness. Dizygotic twins bearing the same hemizygous mutation showed a different retinal phenotype in regard to the severity of the symptoms. This is the first RPGR mutation screening in Poland showing a prevalence of 18% of RPGR pathogenic mutations and likely pathogenic variants in the studied cohort of male patients with an RP phenotype.


Subject(s)
Eye Proteins , Retinitis Pigmentosa , Humans , Male , Child , Adolescent , Young Adult , Adult , Middle Aged , Poland , Eye Proteins/genetics , Pedigree , Phenotype , Retinitis Pigmentosa/pathology
17.
Cell Rep ; 42(9): 113054, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37656622

ABSTRACT

Most mutations in retinitis pigmentosa (RP) arise in rod photoreceptors, but cone photoreceptors, responsible for high-resolution daylight and color vision, are subsequently affected, causing the most debilitating features of the disease. We used mass spectroscopy to follow 13C metabolites delivered to the outer retina and single-cell RNA sequencing to assess photoreceptor transcriptomes. The S cone metabolic transcriptome suggests engagement of the TCA cycle and ongoing response to ROS characteristic of oxidative phosphorylation, which we link to their histone modification transcriptome. Tumor necrosis factor (TNF) and its downstream effector RIP3, which drive ROS generation via mitochondrial dysfunction, are induced and activated as S cones undergo early apoptosis in RP. The long/medium-wavelength (L/M) cone transcriptome shows enhanced glycolytic capacity, which maintains their function as RP progresses. Then, as extracellular glucose eventually diminishes, L/M cones are sustained in long-term dormancy by lactate metabolism.


Subject(s)
Retinal Cone Photoreceptor Cells , Retinitis Pigmentosa , Humans , Retinal Cone Photoreceptor Cells/metabolism , Transcriptome/genetics , Reactive Oxygen Species/metabolism , Retina/metabolism , Retinitis Pigmentosa/pathology
18.
Arch Soc Esp Oftalmol (Engl Ed) ; 98(11): 646-655, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37640142

ABSTRACT

Retinitis pigmentosa (RP) is a group of inherited diseases that lead to degeneration of the retina and decreased vision. The World Health Organization reports around 1,300 million people affected by some type of visual impairment worldwide. The prevalence is 1 in every 4000 inhabitants and it is the first cause of blindness of genetic origin, frequent in men with a percentage of 60% and 40% in women. There is a lack of information on this pathology in the world, mainly on the existing treatments for this disease, so this bibliographic review aims to update the existing or under-study treatments and inform the limitations of each of these therapies. This review of scientific literature was carried out by consulting databases such as PubMed and Web of science, the search will be limited to articles from the years 2018-2022. There are several types of therapy in studies: gene therapy, transcorneal electrical stimulation, use of neuroprotectors, optogenic therapy, stem cell transplants and oligonucleotide therapy, which will be discussed in this article, both their benefits and the existing barriers in each treatment experimental. In conclusion, each of these therapies promises a viable treatment in the future for selective groups of people with retinitis pigmentosa, however, some therapies have shown benefit at the beginning of the disease, losing their efficacy in the long term.


Subject(s)
Retinitis Pigmentosa , Male , Humans , Female , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/pathology , Retina/pathology , Blindness , Genetic Therapy
19.
Stem Cells Dev ; 32(21-22): 681-692, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37470211

ABSTRACT

Retinitis pigmentosa (RP) is a prevalent inherited retinal degenerative disease resulting from photoreceptor and pigment epithelial apoptosis. The Rhodopsin (RHO) is the most commonly associated pathogenic gene in RP. However, RHO mutations (c.512C>T P171L) have been infrequently reported, and the RP pathogenesis caused by these mutations remains unclear. The objective of this study was to investigate the impact of RHO (c.512C>T P171L) mutation on retinal cell differentiation and elucidate the underlying mechanisms of RP. An effective retinal organoid induction scheme for inhibiting the Wnt signaling pathway was selected for further experiments, and the established cell line chHES-406 was demonstrated to be heterozygous for RHO c.512C>T, with a normal karyotype and pluripotency potential. Furthermore, the development of chHES-406 organoids may be delayed, and apoptosis detection and co-localization revealed that chHES-406 organoids had more apoptotic cells than chHES-90 in the outer nuclear layer (ONL), mutant RHO protein was mislocalized in the endoplasmic reticulum (ER), and stress-related and apoptotic gene expression increased. Overall, our study elucidated a possible mechanism by which ER stress caused by RHO P171L protein mislocalization may lead to ONL cell apoptosis.


Subject(s)
Retinitis Pigmentosa , Rhodopsin , Humans , Rhodopsin/genetics , Rhodopsin/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Endoplasmic Reticulum Stress/genetics , Apoptosis/genetics , Mutation/genetics
20.
Adv Exp Med Biol ; 1415: 97-102, 2023.
Article in English | MEDLINE | ID: mdl-37440020

ABSTRACT

Mutations in peripherin 2 (PRPH2) are associated with a spectrum of inherited retinal diseases (IRDs) including retinitis pigmentosa (RP) and macular degeneration. As PRPH2 is localized to cone and rod outer segments, mutations in PRPH2 lead the disorganization or absence of photoreceptor outer segments. Here, we report on a patient with PRPH2-linked RP who exhibited widespread RPE atrophy with a central area of macular atrophy sparing the fovea. In future studies, we plan to model the pathobiology of PRPH2-based RP using induced pluripotent stem cell (iPSC)-derived retinal organoids. To effectively model rare mutations using iPSC-derived retinal organoids, we first require a strategy that can install the desired mutation in healthy wild-type iPSC, which can efficiently generate well-laminated retinal organoids. In this study, we developed an efficient prime editing strategy for the installation of the pathogenic PRPH2 c.828+1 G>A splice-site mutation underlying our patient's disease.


Subject(s)
Macular Degeneration , Retinal Degeneration , Retinitis Pigmentosa , Humans , Peripherins/genetics , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Mutation , Atrophy
SELECTION OF CITATIONS
SEARCH DETAIL
...